Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Publication year range
1.
Cell ; 185(20): 3705-3719.e14, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179667

ABSTRACT

The intestinal microbiota is an important modulator of graft-versus-host disease (GVHD), which often complicates allogeneic hematopoietic stem cell transplantation (allo-HSCT). Broad-spectrum antibiotics such as carbapenems increase the risk for intestinal GVHD, but mechanisms are not well understood. In this study, we found that treatment with meropenem, a commonly used carbapenem, aggravates colonic GVHD in mice via the expansion of Bacteroides thetaiotaomicron (BT). BT has a broad ability to degrade dietary polysaccharides and host mucin glycans. BT in meropenem-treated allogeneic mice demonstrated upregulated expression of enzymes involved in the degradation of mucin glycans. These mice also had thinning of the colonic mucus layer and decreased levels of xylose in colonic luminal contents. Interestingly, oral xylose supplementation significantly prevented thinning of the colonic mucus layer in meropenem-treated mice. Specific nutritional supplementation strategies, including xylose supplementation, may combat antibiotic-mediated microbiome injury to reduce the risk for intestinal GVHD in allo-HSCT patients.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteroides , Carbapenems/pharmacology , Carbapenems/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Meropenem , Mice , Mucins/metabolism , Mucus/metabolism , Polysaccharides/metabolism , Xylose
2.
Cytotherapy ; 23(9): 793-798, 2021 09.
Article in English | MEDLINE | ID: mdl-34215503

ABSTRACT

BACKGROUND AIMS: Human myeloperoxidase has been shown to be overexpressed in many types of leukemia, such as chronic myeloid leukemia, acute myeloid leukemia and myelodysplastic syndrome. The authors identified two myeloperoxidase-derived HLA-A2-restricted peptides, MY4 and MY8, as novel leukemia-associated antigens. METHODS: Ex vivo-elicited MY4- and MY8-specific cytotoxic T lymphocytes were generated, and tested for leukemia cell lysis in vitro and in NOD/SCID AML xenograft model. RESULTS: These MY4- and MY8-specific cytotoxic T lymphocytes killed leukemic blasts while sparing healthy donor bone marrow cells. In addition, co-injection of MY4- and MY8-specific cytotoxic T lymphocytes into nonobese diabetic/severe combined immunodeficiency mice with acute myeloid leukemia drastically reduced tumor burden in vivo. The authors also found that MY4- and MY8-specific T cells could be detected in the peripheral blood mononuclear cells of allogeneic stem cell transplant recipients. CONCLUSIONS: These antigen-specific T cells were significantly increased in blood samples from patients compared with healthy donors, suggesting that both MY4 and MY8 are immunogenic and that MY4- and MY8-specific cytotoxic T lymphocytes may play a role in reducing leukemia in vivo. Thus, the discovery of MY4 and MY8 as novel leukemia-associated antigens paves the way for targeting these antigens in immunotherapy against myeloid leukemia.


Subject(s)
HLA-A2 Antigen , Leukemia, Myeloid, Acute , Animals , Humans , Leukemia, Myeloid, Acute/therapy , Leukocytes, Mononuclear , Mice , Mice, Inbred NOD , Mice, SCID , Peptides , Peroxidase , T-Lymphocytes, Cytotoxic
3.
J Immunol ; 201(5): 1389-1399, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30021768

ABSTRACT

Proteinase 3 (P3), a serine protease expressed by myeloid cells, localized within azurophil granules, and also expressed on the cellular membrane of polymorphonuclear neutrophils (PMN), is the target of autoimmunity in granulomatosis with polyangiitis. PR1, an HLA-A2 restricted nonameric peptide derived from P3, has been targeted effectively in myeloid leukemia. We previously showed (Molldrem et al. 2003. JClinInvest 111: 639-647) that overexpression of P3 in chronic myeloid leukemia induces apoptosis of high-affinity PR1-specific T cells, leading to deletional tolerance and leukemia outgrowth. In this study, we investigated the effect of membrane P3 (mP3)-expressing PMN and acute myeloid leukemia (AML) blasts on the proliferation of CD4 and CD8 T cells in vitro. We demonstrate that mP3-expressing PMN significantly inhibits autologous healthy donor T cell proliferation but does not affect cytokine production in activated T cells and that this effect requires cell proximity and was abrogated by P3 blockade. This inhibition required P3 enzyme activity. However, suppression was not reversed by either the addition of catalase or the inhibition of arginase I. In addition to P3 blockade, anti-low density lipoprotein receptor-related protein 1 (LRP1) Ab also restored T cells' capacity to proliferate. Last, we show dose-dependent inhibition of T cell proliferation by mP3-expressing AML blasts. Together, our findings demonstrate a novel mechanism whereby PMN- and AML-associated mP3 inhibits T cell proliferation via direct LRP1 and mP3 interaction, and we identify P3 as a novel target to modulate immunity in myeloid leukemia and autoimmune disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Leukemia, Myeloid, Acute/immunology , Myeloblastin/immunology , Neoplasm Proteins/immunology , Neutrophils/immunology , Adult , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Neutrophils/pathology
4.
Biol Blood Marrow Transplant ; 25(7): 1347-1354, 2019 07.
Article in English | MEDLINE | ID: mdl-30826465

ABSTRACT

Although bortezomib and rituximab have synergistic activity in patients with lymphoma and both can attenuate graft-versus-host disease (GVHD), the drugs have not been used together in patients undergoing allogeneic stem cell transplantation (alloSCT). In this phase I/II trial, we assessed the safety and activity of bortezomib added to the rituximab (R) plus BEAM (carmustine, etoposide, cytarabine, melphalan) regimen in patients with relapsed lymphoma undergoing alloSCT. Primary GVHD prophylaxis consisted of tacrolimus and methotrexate. Bortezomib (1 to 1.3 mg/m2 per dose) was administered i.v. on days -13, -6, -1, and +2. We performed inverse probability weighting analysis to compare GVHD and survival results with an historical control group that received R-BEAM without bortezomib. Thirty-nine patients were assessable for toxic effects and response. The median age was 54 years. The most common diagnosis was diffuse large B cell lymphoma (41%). Twenty-two patients (56%) and 17 patients (44%) received their transplants from matched related and matched unrelated donors, respectively. The maximum tolerated bortezomib dose was 1 mg/m2. The weighted cumulative incidences of grades II to IV and III or IV acute GVHD were 50% and 34%, respectively; these incidences and survival rates were not significantly different from those of the control group. Median survival was not reached in patients age ≤ 50 years and with a long follow-up time of 60.7 months. The R-BEAM regimen has a survival benefit in lymphoma patients age ≤ 50 years undergoing alloSCT. The addition of bortezomib has no impact on survival or incidence of GVHD.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bortezomib/administration & dosage , Hematologic Neoplasms , Lymphoma, Large B-Cell, Diffuse , Rituximab/administration & dosage , Stem Cell Transplantation , Adult , Age Factors , Aged , Allografts , Carmustine/administration & dosage , Cytarabine/administration & dosage , Disease-Free Survival , Female , Hematologic Neoplasms/mortality , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Male , Melphalan/administration & dosage , Middle Aged , Podophyllotoxin/administration & dosage , Survival Rate
5.
Br J Haematol ; 185(4): 679-690, 2019 05.
Article in English | MEDLINE | ID: mdl-30828801

ABSTRACT

Acute myeloid leukaemia (AML) is a lethal haematological malignancy characterized by an immunosuppressive milieu in the tumour microenvironment (TME) that fosters disease growth and therapeutic resistance. Hypomethylating agents (HMAs) demonstrate clinical efficacy in AML patients and exert immunomodulatory activities. In the present study, we show that guadecitabine augments both antigen processing and presentation, resulting in increased AML susceptibility to T cell-mediated killing. Exposure to HMA results in the activation of the endogenous retroviral pathway with concomitant downstream amplification of critical mediators of inflammation. In an immunocompetent murine leukaemia model, guadecitabine negatively regulates inhibitory accessory cells in the TME by decreasing PD-1 (also termed PDCD1) expressing T cells and reducing AML-mediated expansion of myeloid-derived suppressor cells. Therapy with guadecitabine results in enhanced leukaemia-specific immunity, as manifested by increased CD4 and CD8 cells targeting syngeneic leukaemia cells. We have previously reported that vaccination with AML/dendritic cell fusions elicits the expansion of leukaemia-specific T cells and protects against disease relapse. In the present study, we demonstrate that vaccination in conjunction with HMA therapy results in enhanced anti-leukaemia immunity and survival. The combination of a novel personalized dendritic cell/AML fusion vaccine and an HMA has therapeutic potential, and a clinical trial investigating this combination is planned.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Azacitidine/analogs & derivatives , Cancer Vaccines/immunology , Leukemia, Myeloid, Acute/drug therapy , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents, Immunological/immunology , Azacitidine/immunology , Azacitidine/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/immunology , Humans , Immunity, Cellular/drug effects , Leukemia, Myeloid, Acute/immunology , Mice, Inbred C57BL , Neoplasm Transplantation , Programmed Cell Death 1 Receptor/metabolism , Retroviridae/immunology , Virus Activation/immunology
6.
Proc Natl Acad Sci U S A ; 113(11): E1555-64, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929325

ABSTRACT

The immunoproteasome plays a key role in generation of HLA peptides for T cell-mediated immunity. Integrative genomic and proteomic analysis of non-small cell lung carcinoma (NSCLC) cell lines revealed significantly reduced expression of immunoproteasome components and their regulators associated with epithelial to mesenchymal transition. Low expression of immunoproteasome subunits in early stage NSCLC patients was associated with recurrence and metastasis. Depleted repertoire of HLA class I-bound peptides in mesenchymal cells deficient in immunoproteasome components was restored with either IFNγ or 5-aza-2'-deoxycytidine (5-aza-dC) treatment. Our findings point to a mechanism of immune evasion of cells with a mesenchymal phenotype and suggest a strategy to overcome immune evasion through induction of the immunoproteasome to increase the cellular repertoire of HLA class I-bound peptides.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Proteasome Endopeptidase Complex/genetics , Adult , Aged , Aged, 80 and over , Antigens, CD/immunology , Antigens, CD/metabolism , Cadherins/immunology , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , HLA Antigens/metabolism , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Proteasome Endopeptidase Complex/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology
7.
J Biol Chem ; 292(24): 10295-10305, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28468826

ABSTRACT

Neutrophil elastase (NE) can be rapidly taken up by tumor cells that lack endogenous NE expression, including breast cancer, which results in cross-presentation of PR1, an NE-derived HLA-A2-restricted peptide that is an immunotherapy target in hematological and solid tumor malignancies. The mechanism of NE uptake, however, remains unknown. Using the mass spectrometry-based approach, we identify neuropilin-1 (NRP1) as a NE receptor that mediates uptake and PR1 cross-presentation in breast cancer cells. We demonstrated that soluble NE is a specific, high-affinity ligand for NRP1 with a calculated Kd of 38.7 nm Furthermore, we showed that NRP1 binds to the RRXR motif in NE. Notably, NRP1 knockdown with interfering RNA or CRISPR-cas9 system and blocking using anti-NRP1 antibody decreased NE uptake and, subsequently, susceptibility to lysis by PR1-specific cytotoxic T cells. Expression of NRP1 in NRP1-deficient cells was sufficient to induce NE uptake. Altogether, because NRP1 is broadly expressed in tumors, our findings suggest a role for this receptor in immunotherapy strategies that target cross-presented antigens.


Subject(s)
Absorption, Physiological , Breast Neoplasms/metabolism , Cross-Priming , Leukocyte Elastase/metabolism , Neoplasm Proteins/metabolism , Neuropilin-1/metabolism , Amino Acid Motifs , Antibodies, Blocking/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Line, Tumor , Female , Humans , Kinetics , Leukocyte Elastase/chemistry , Leukocyte Elastase/immunology , Ligands , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/chemistry , Neuropilin-1/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
8.
Blood ; 125(19): 2885-92, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25778529

ABSTRACT

Delayed engraftment is a major limitation of cord blood transplantation (CBT), due in part to a defect in the cord blood (CB) cells' ability to home to the bone marrow. Because this defect appears related to low levels of fucosylation of cell surface molecules that are responsible for binding to P- and E-selectins constitutively expressed by the marrow microvasculature, and thus for marrow homing, we conducted a first-in-humans clinical trial to correct this deficiency. Patients with high-risk hematologic malignancies received myeloablative therapy followed by transplantation with 2 CB units, one of which was treated ex vivo for 30 minutes with the enzyme fucosyltransferase-VI and guanosine diphosphate fucose to enhance the interaction of CD34(+) stem and early progenitor cells with microvessels. The results of enforced fucosylation for 22 patients enrolled in the trial were then compared with those for 31 historical controls who had undergone double unmanipulated CBT. The median time to neutrophil engraftment was 17 days (range, 12-34 days) compared with 26 days (range, 11-48 days) for controls (P = .0023). Platelet engraftment was also improved: median was 35 days (range, 18-100 days) compared with 45 days (range, 27-120 days) for controls (P = .0520). These findings support ex vivo fucosylation of multipotent CD34(+) CB cells as a clinically feasible means to improve engraftment efficiency in the double CBT setting. The trial is registered to www.clinicaltrials.gov as #NCT01471067.


Subject(s)
Blood Platelets/cytology , Fetal Blood/cytology , Fucose/metabolism , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Neutrophils/transplantation , Adolescent , Adult , Aged , Blood Platelets/immunology , Cohort Studies , E-Selectin/metabolism , Feasibility Studies , Female , Fetal Blood/immunology , Fucosyltransferases/metabolism , Graft vs Host Disease , Hematologic Neoplasms/immunology , Hematologic Neoplasms/mortality , Hematopoietic Stem Cells/immunology , Humans , Male , Middle Aged , Neutrophils/cytology , Neutrophils/immunology , P-Selectin/metabolism , Platelet Transfusion , Prognosis , Survival Rate , Young Adult
9.
Cancer Immunol Immunother ; 65(6): 741-51, 2016 06.
Article in English | MEDLINE | ID: mdl-27129972

ABSTRACT

Neutrophil elastase (NE) is an innate immune cell-derived inflammatory mediator that we have shown increases the presentation of tumor-associated peptide antigens in breast cancer. In this study, we extend these observations to show that NE uptake has a broad effect on enhancing antigen presentation by breast cancer cells. We show that NE increases human leukocyte antigen (HLA) class I expression on the surface of breast cancer cells in a concentration and time-dependent manner. HLA class I upregulation requires internalization of enzymatically active NE. Western blots of NE-treated breast cancer cells confirm that the expression of total HLA class I as well as the antigen-processing machinery proteins TAP1, LMP2, and calnexin does not change following NE treatment. This suggests that NE does not increase the efficiency of antigen processing; rather, it mediates the upregulation of HLA class I by stabilizing and reducing membrane recycling of HLA class I molecules. Furthermore, the effects of NE extend beyond breast cancer since the uptake of NE by EBV-LCL increases the presentation of HLA class I-restricted viral peptides, as shown by their increased sensitivity to lysis by EBV-specific CD8+ T cells. Together, our results show that NE uptake increases the responsiveness of breast cancer cells to adaptive immunity by broad upregulation of membrane HLA class I and support the conclusion that the innate inflammatory mediator NE enhances tumor cell recognition and increases tumor sensitivity to the host adaptive immune response.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Leukocyte Elastase/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Humans , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
10.
Blood ; 123(25): 3849-50, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24948621

ABSTRACT

In this issue of Blood, Zhou et al report long-term follow-up and detailed analysis of immune reconstitution associated with a different suicide gene strategy to abrogate graft-versus-host disease (GVHD).


Subject(s)
Caspase 9/genetics , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/transplantation , Transgenes/genetics , Female , Humans , Male
11.
Cytotherapy ; 18(8): 985-994, 2016 08.
Article in English | MEDLINE | ID: mdl-27265873

ABSTRACT

BACKGROUND AIMS: The PR1 peptide, derived from the leukemia-associated antigens proteinase 3 and neutrophil elastase, is overexpressed on HLA-A2 in acute myeloid leukemia (AML). We developed a T-cell receptor (TCR)-like monoclonal antibody (8F4) that binds the PR1/HLA-A2 complex on the surface of AML cells, efficiently killing them in vitro and eliminating them in preclinical models. Humanized 8F4 (h8F4) with high affinity for the PR1/HLA-A2 epitope was used to construct an h8F4- chimeric antigen receptor (CAR) that was transduced into T cells to mediate anti-leukemia activity. METHODS: Human T cells were transduced to express the PR1/HLA-A2-specific CAR (h8F4-CAR-T cells) containing the scFv of h8F4 fused to the intracellular signaling endo-domain of CD3 zeta chain through the transmembrane and intracellular costimulatory domain of CD28. RESULTS: Adult human normal peripheral blood (PB) T cells were efficiently transduced with the h8F4-CAR construct and predominantly displayed an effector memory phenotype with a minor population (12%) of central memory cells in vitro. Umbilical cord blood (UCB) T cells could also be efficiently transduced with the h8F4-CAR. The PB and UCB-derived h8F4-CAR-T cells specifically recognized the PR1/HLA-A2 complex and were capable of killing leukemia cell lines and primary AML blasts in an HLA-A2-dependent manner. CONCLUSIONS: Human adult PB and UCB-derived T cells expressing a CAR derived from the TCR-like 8F4 antibody rapidly and efficiently kill AML in vitro. Our data could lead to a new treatment paradigm for AML in which targeting leukemia stem cells could transfer long-term immunity to protect against relapse.


Subject(s)
Fetal Blood , HLA-A2 Antigen/immunology , Leukemia, Myeloid, Acute/therapy , Leukocytes, Mononuclear/metabolism , Myeloblastin/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Adult , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cell Line , Epitopes/immunology , Fetal Blood/cytology , Fetal Blood/immunology , Genetic Therapy , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/immunology , Myeloblastin/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocytes/immunology
12.
Cytotherapy ; 18(8): 995-1001, 2016 08.
Article in English | MEDLINE | ID: mdl-27378343

ABSTRACT

BACKGROUND AIMS: PR1 is an HLA-A2 restricted leukemia-associated antigen derived from neutrophil elastase and proteinase 3, both of which are normally stored in the azurophil granules of myeloid cells but overexpressed in myeloid leukemic cells. PR1-specific cytotoxic lymphocytes (PR1-CTLs) have activity against primary myeloid leukemia in vitro and in vivo and thus could have great potential in the setting of adoptive cellular therapy (ACT). Adult peripheral blood-derived PR1-CTLs are infrequent but preferentially lyse myeloid leukemia cells. We sought to examine PR1-CTLs in umbilical cord blood (UCB) because UCB units provide a rapidly available cell source and a lower risk of graft-versus-host disease, even in the setting of mismatched human leukocyte antigen (HLA) loci. METHODS: We first determined the frequency of PR1-CTLs in HLA-A2(+) UCB units and then successfully expanded them ex vivo using repeated stimulation with PR1 peptide-pulsed antigen-presenting cells (APCs). After expansion, we assessed the PR1-CTL phenotype (naive, effector, memory) and function against PR1-expressing target cells. RESULTS: PR1-CTLs are detected at an average frequency of 0.14% within the CD8(+) population of fresh UCB units, which is 45 times higher than in healthy adult peripheral blood. UCB PR1-CTLs are phenotypically naive, consistent with the UCB CD8(+) population as a whole. In addition, the cells can be expanded by stimulation with PR1 peptide-pulsed APCs. Expansion results in an increased frequency of PR1-CTLs, up to 4.56%, with an average 20-fold increase in total number. After expansion, UCB PR1-CTLs express markers consistent with effector memory T cells. Expanded UCB PR1-CTLs are functional in vitro as they are able to produce cytokines and lyse PR1-expressing leukemia cell lines. CONCLUSIONS: This study is the first report to show that T cells specific for a leukemia-associated antigen are found at a significantly higher frequency in UCB than adult blood. Our results also demonstrate specific cytotoxicity of expanded UCB-derived PR1-CTLs against PR1-expressing targets. Together, our data suggest that UCB PR1-CTLs could be useful to prevent or treat leukemia relapse in myeloid leukemia patients.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Fetal Blood/cytology , HLA-A2 Antigen/immunology , Immunotherapy, Adoptive , Leukemia, Myeloid/therapy , Myeloblastin/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Cells, Cultured , Fetal Blood/immunology , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/metabolism , Humans , K562 Cells , Leukemia, Myeloid/immunology , Lymphocyte Count , Myeloblastin/chemistry , Myeloblastin/metabolism , T-Lymphocytes, Cytotoxic/metabolism , U937 Cells
13.
Proc Natl Acad Sci U S A ; 110(51): 20717-22, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297924

ABSTRACT

Angiogenesis is fundamental to tumorigenesis and an attractive target for therapeutic intervention against cancer. We have recently demonstrated that CD13 (aminopeptidase N) expressed by nonmalignant host cells of unspecified types regulate tumor blood vessel development. Here, we compare CD13 wild-type and null bone marrow-transplanted tumor-bearing mice to show that host CD13(+) bone marrow-derived cells promote cancer progression via their effect on angiogenesis. Furthermore, we have identified CD11b(+)CD13(+) myeloid cells as the immune subpopulation directly regulating tumor blood vessel development. Finally, we show that these cells are specifically localized within the tumor microenvironment and produce proangiogenic soluble factors. Thus, CD11b(+)CD13(+) myeloid cells constitute a population of bone marrow-derived cells that promote tumor progression and metastasis and are potential candidates for the development of targeted antiangiogenic drugs.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Bone Marrow Cells/metabolism , CD13 Antigens , Myeloid Cells/metabolism , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Animals , Bone Marrow Cells/pathology , CD11b Antigen , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/pathology , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/pathology
14.
N Engl J Med ; 367(24): 2305-15, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23234514

ABSTRACT

BACKGROUND: Poor engraftment due to low cell doses restricts the usefulness of umbilical-cord-blood transplantation. We hypothesized that engraftment would be improved by transplanting cord blood that was expanded ex vivo with mesenchymal stromal cells. METHODS: We studied engraftment results in 31 adults with hematologic cancers who received transplants of 2 cord-blood units, 1 of which contained cord blood that was expanded ex vivo in cocultures with allogeneic mesenchymal stromal cells. The results in these patients were compared with those in 80 historical controls who received 2 units of unmanipulated cord blood. RESULTS: Coculture with mesenchymal stromal cells led to an expansion of total nucleated cells by a median factor of 12.2 and of CD34+ cells by a median factor of 30.1. With transplantation of 1 unit each of expanded and unmanipulated cord blood, patients received a median of 8.34×10(7) total nucleated cells per kilogram of body weight and 1.81×10(6) CD34+ cells per kilogram--doses higher than in our previous transplantations of 2 units of unmanipulated cord blood. In patients in whom engraftment occurred, the median time to neutrophil engraftment was 15 days in the recipients of expanded cord blood, as compared with 24 days in controls who received unmanipulated cord blood only (P<0.001); the median time to platelet engraftment was 42 days and 49 days, respectively (P=0.03). On day 26, the cumulative incidence of neutrophil engraftment was 88% with expansion versus 53% without expansion (P<0.001); on day 60, the cumulative incidence of platelet engraftment was 71% and 31%, respectively (P<0.001). CONCLUSIONS: Transplantation of cord-blood cells expanded with mesenchymal stromal cells appeared to be safe and effective. Expanded cord blood in combination with unmanipulated cord blood significantly improved engraftment, as compared with unmanipulated cord blood only. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT00498316.).


Subject(s)
Cord Blood Stem Cell Transplantation , Hematologic Neoplasms/therapy , Mesenchymal Stem Cell Transplantation , Adolescent , Adult , Blood Cell Count , Blood Platelets , Cause of Death , Cell Culture Techniques , Graft Enhancement, Immunologic , Graft vs Host Disease , Hematologic Neoplasms/mortality , Humans , Mesenchymal Stem Cells , Middle Aged , Neutrophils , Transplantation Chimera , Transplantation, Homologous , Young Adult
15.
Genet Epidemiol ; 37(3): 267-75, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23471868

ABSTRACT

Joint testing for the cumulative effect of multiple single-nucleotide polymorphisms grouped on the basis of prior biological knowledge has become a popular and powerful strategy for the analysis of large-scale genetic association studies. The kernel machine (KM)-testing framework is a useful approach that has been proposed for testing associations between multiple genetic variants and many different types of complex traits by comparing pairwise similarity in phenotype between subjects to pairwise similarity in genotype, with similarity in genotype defined via a kernel function. An advantage of the KM framework is its flexibility: choosing different kernel functions allows for different assumptions concerning the underlying model and can allow for improved power. In practice, it is difficult to know which kernel to use a priori because this depends on the unknown underlying trait architecture and selecting the kernel which gives the lowest P-value can lead to inflated type I error. Therefore, we propose practical strategies for KM testing when multiple candidate kernels are present based on constructing composite kernels and based on efficient perturbation procedures. We demonstrate through simulations and real data applications that the procedures protect the type I error rate and can lead to substantially improved power over poor choices of kernels and only modest differences in power vs. using the best candidate kernel.


Subject(s)
Models, Genetic , Polymorphism, Single Nucleotide , Premature Birth/genetics , Software , Computer Simulation , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Infant, Newborn , Phenotype , Pregnancy
16.
Blood ; 120(8): 1540-1, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22918422

ABSTRACT

The Wilms tumor protein, WT-1, is a widely recognized tumor antigen that is aberrantly expressed in myeloid and lymphoid leukemia and in this issue of Blood, Doubrovina et al report the most extensive catalog heretofore of HLA-restricted immunogenic peptides derived from WT-1, which are recognized by CD8 and CD4T cells.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Leukemia/immunology , Peptides/immunology , T-Lymphocytes/immunology , WT1 Proteins/immunology , Humans
18.
J Immunol ; 189(11): 5476-84, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23105141

ABSTRACT

PR1 is a HLA-A2-restricted peptide that has been targeted successfully in myeloid leukemia with immunotherapy. PR1 is derived from the neutrophil granule proteases proteinase 3 (P3) and neutrophil elastase (NE), which are both found in the tumor microenvironment. We recently showed that P3 and NE are taken up and cross-presented by normal and leukemia-derived APCs, and that NE is taken up by breast cancer cells. We now extend our findings to show that P3 and NE are taken up and cross-presented by human solid tumors. We further show that PR1 cross-presentation renders human breast cancer and melanoma cells susceptible to killing by PR1-specific CTLs (PR1-CTL) and the anti-PR1/HLA-A2 Ab 8F4. We also show PR1-CTL in peripheral blood from patients with breast cancer and melanoma. Together, our data identify cross-presentation as a novel mechanism through which cells that lack endogenous expression of an Ag become susceptible to therapies that target cross-presented Ags and suggest PR1 as a broadly expressed tumor Ag.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/therapy , Immunotherapy , Leukocyte Elastase/immunology , Melanoma/therapy , Myeloblastin/immunology , Skin Neoplasms/therapy , Antibodies/pharmacology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cross-Priming , Female , HLA-A2 Antigen/immunology , Humans , Leukocyte Elastase/chemistry , Melanoma/immunology , Melanoma/pathology , Molecular Targeted Therapy , Myeloblastin/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
19.
Proc Natl Acad Sci U S A ; 108(46): 18637-42, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22049339

ABSTRACT

Molecules differentially expressed in blood vessels among organs or between damaged and normal tissues, are attractive therapy targets; however, their identification within the human vasculature is challenging. Here we screened a peptide library in cancer patients to uncover ligand-receptors common or specific to certain vascular beds. Surveying ~2.35 x 10(6) motifs recovered from biopsies yielded a nonrandom distribution, indicating that systemic tissue targeting is feasible. High-throughput analysis by similarity search, protein arrays, and affinity chromatography revealed four native ligand-receptors, three of which were previously unrecognized. Two are shared among multiple tissues (integrin α4/annexin A4 and cathepsin B/apolipoprotein E3) and the other two have a restricted and specific distribution in normal tissue (prohibitin/annexin A2 in white adipose tissue) or cancer (RAGE/leukocyte proteinase-3 in bone metastases). These findings provide vascular molecular markers for biotechnology and medical applications.


Subject(s)
Blood Vessels/metabolism , Bone Marrow/metabolism , Neoplasms/metabolism , Amino Acid Motifs , Annexin A4/biosynthesis , Apolipoprotein E3/biosynthesis , Biopsy , Cathepsin B/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha4/biosynthesis , Ligands , Neovascularization, Pathologic , Obesity/metabolism , Peptide Library
20.
Cancer Res Commun ; 4(3): 911-918, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38477596

ABSTRACT

Lack of robust activation of Stimulator of Interferon Genes (STING) pathway and subsequent induction of type I IFN responses is considered a barrier to antitumor immunity in acute myeloid leukemia (AML). Using common human AML cell lines as in vitro tools to evaluate the efficacy of novel STING agonists, we found most AML lines to be poor producers of IFNs upon exposure to extremely potent agonists, suggesting cell-intrinsic suppression of STING signaling may occur. We observed unexpected patterns of response that did not correlate with levels of STING pathway components or of known enzymes associated with resistance. To identify a genetic basis for these observations, we cloned and sequenced STING from the cDNA of human AML cell lines and found both frequent mutations and deviations from normal RNA splicing. We identified two novel spliced isoforms of STING in these lines and validated their expression in primary human AML samples. When transduced into reporter cells, these novel STING isoforms exhibited complete insensitivity to agonist stimulation. These observations identify alternative splicing as a mechanism of STING pathway suppression and suggest that most AML silences the STING pathway through direct modification rather than through engagement of external inhibitory factors. SIGNIFICANCE: We find that AML acquires resistance to innate immune activation via the STING pathway through aberrant splicing of the STING transcript including two novel forms described herein that act as dominant negatives. These data broaden understanding of how cancers evolve STING resistance, and suggest that the AML tumor microenvironment, not the cancer cell, should be the target of therapeutic interventions to activate STING.


Subject(s)
Interferon Type I , Leukemia, Myeloid, Acute , Humans , Protein Isoforms/genetics , Leukemia, Myeloid, Acute/genetics , Alternative Splicing/genetics , Interferon Type I/genetics , Cell Line , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL